1
|
Li Y, Wang X, Chen K, Zhuang Z, Tang H, Yu T, Cao W. Efficient production of 2'-fucosyllactose in Pichia pastoris through metabolic engineering and constructing an orthogonal energy supply system. Synth Syst Biotechnol 2025; 10:807-815. [PMID: 40297761 PMCID: PMC12035725 DOI: 10.1016/j.synbio.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/30/2025] [Accepted: 04/03/2025] [Indexed: 04/30/2025] Open
Abstract
2'-fucosyllactose (2'-FL) holds significant role in the infants' nutrition. While microbial production of 2'-FL has predominantly utilized Escherichia coli and Saccharomyces cerevisiae, the potential of Pichia pastoris, renowned for its robust NADPH regeneration capability, remains underexplored. Herein, we systematically engineered the metabolism of P. pastoris to develop an efficient 2'-FL-producing cell factory. We first constructed the de novo biosynthesis pathway for 2'-FL in P. pastoris, achieving an initial titer of 0.143 g/L. By optimizing enzyme selection and solubility of α-1,2-fucosyltransferase (FutC), 2'-FL production was enhanced by nearly ten folds. Subsequently, engineering NADPH supply further increased the 2'-FL production by 170 %. Furthermore, we enhanced energy supply by incorporating an orthogonal energy module based on the methanol dissimilation pathway and increasing GTP availability, resulting in a 32 % improvement in 2'-FL production. Finally, through the optimization of fermentation condition, we realized the production titer of 2'-FL to 3.50 g/L in shake-flask, representing the highest titer in P. pastoris. These findings highlight the potential of P. pastoris as a chassis to produce chemicals by providing abundant NADPH and utilizing methanol as co-substrate to supply sufficient energy.
Collapse
Affiliation(s)
- Yi Li
- Center for Synthetic Biochemistry, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiang Wang
- Center for Synthetic Biochemistry, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kaidi Chen
- Center for Synthetic Biochemistry, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhoukang Zhuang
- Center for Synthetic Biochemistry, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen, 518055, China
| | - Hongting Tang
- School of Agriculture and Biotechnology, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518107, China
| | - Tao Yu
- Center for Synthetic Biochemistry, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen, 518055, China
| | - Wenbing Cao
- Center for Synthetic Biochemistry, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen, 518055, China
| |
Collapse
|
2
|
Ito K, Islam J, Sakurai K, Koyama S, Matsuo A, Okano K, Hirakawa R, Furukawa M, Nochi T. Breast milk stabilizes bacterial communities in the large intestine even after weaning. Biochem Biophys Res Commun 2025; 756:151585. [PMID: 40068432 DOI: 10.1016/j.bbrc.2025.151585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 03/22/2025]
Abstract
The development and maintenance of a balanced microbiota is crucial for human health. Milk contains immune factors that not only protect offspring from infectious diseases but also play an important role in promoting the development and maintenance of the microbiota. However, the persisting effects of milk-derived immune factors on the maintenance of the microbiota after weaning have not been carefully examined. In this study, a cross-fostering model was employed using immunocompetent (IC) and immunodeficient (ID) mice in which one-half of the pups born from two dams were replaced. As a result, breast milk from the IC dam (IC milk) affected the development of the microbiota during lactation and maintained it even after weaning in the large intestine of the ID pups. The large intestinal microbiota of ID pups raised on IC milk remained similar to that of normal IC pups. Under normal conditions, the genus Mucispirillum was closely associated with other bacteria, forming a diverse bacterial community in the large intestine. In the small intestine, there were no differences in the microbiota before weaning, regardless of whether IC or ID milk was consumed. By contrast, significant differences were observed in the small intestinal microbiota between IC and ID mice after weaning; however, this was dependent on the immune-related characteristics of offspring (rather than milk-derived immune factors). These results indicate that breast milk plays an important role in the large (not small) intestine of offspring to create and maintain a diverse microbiota with a balanced bacterial network even after weaning.
Collapse
Affiliation(s)
- Kaori Ito
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Jahidul Islam
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Kota Sakurai
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Saeka Koyama
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Graduate Program in Food Science, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | | | - Kunihiro Okano
- GENODAS Co., Ltd. Miyagi, 980-8572, Japan; Department of Biological Environment, Faculty of Bioresource Sciences, Akita Prefectural University, Akita, 010-0195, Japan
| | - Ryota Hirakawa
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Mucosal Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Mutsumi Furukawa
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Mucosal Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Tomonori Nochi
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Graduate Program in Food Science, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Laboratory of Animal Mucosal Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan; Division of Mucosal Vaccines, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan; Department of Animal Bioscience, University of Guelph, Ontario, N1G 2W1, Canada; Center for Professional Development, Institute for Excellence in Higher Education, Tohoku University, Miyagi, 980-8576, Japan.
| |
Collapse
|
3
|
Sanz Morales P, Wijeyesekera A, Robertson MD, Major G, Boulangé CL, Jackson PPJ, Poveda Turrado CG, Gibson GR. An in vitro batch culture study to assess the fermentation of human milk oligosaccharides by faecal microbiota from healthy and irritable bowel syndrome stool donors. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2025; 6:e4. [PMID: 40297306 PMCID: PMC12034501 DOI: 10.1017/gmb.2025.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 04/30/2025]
Abstract
This study explored the effects of different human milk oligosaccharides (HMOs), solely and in combination, on gut microbiota composition and metabolic activity (organic acid production), using anaerobic in vitro batch culture fermenters. The aim was to compare prebiotic effects of HMOs (2'FL, 3'FL, 3'SL, 6'SL, LNT, LNnT, and 1:1 ratio mixes of 2'FL/3'SL and 3'SL/LNT) in faecal samples from irritable bowel syndrome (IBS) donors and healthy controls, and to determine the best-performing HMO in IBS. Fluorescent in situ hybridisation coupled with flow cytometry was utilised to study microbiota changes in major colonic genera, and organic acid production was assessed by gas chromatography. IBS donors had different starting microbial profiles compared to healthy controls and lower levels of organic acids. In response to HMOs, there were alterations in both the control and IBS faecal microbiomes. In IBS donor fermenters, Bifidobacterium, Faecalibacterium, total bacterial numbers, and organic acid production significantly increased post-HMO intervention. When comparing the effect of HMO interventions on the microbiota and organic acid production, a mix of 3'SL/LNT HMOs may be the most promising intervention for IBS patients.
Collapse
Affiliation(s)
| | - Anisha Wijeyesekera
- Department of Food and Nutritional Sciences, The University of Reading, Reading, UK
| | - M. Denise Robertson
- Department of Nutrition, Food and Exercise Sciences, Health & Medical Sciences, University of Surrey, Guildford, UK
| | - Giles Major
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | - Claire L. Boulangé
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | | | | | - Glenn R. Gibson
- Department of Food and Nutritional Sciences, The University of Reading, Reading, UK
| |
Collapse
|
4
|
Liu S, Liu M, Wang Y, Liu X, Ren F, Zhang H. Maillard reaction with α-lactalbumin in infant formula reduces the prebiotic properties of lacto-N-neotetraose. Food Res Int 2025; 204:115875. [PMID: 39986755 DOI: 10.1016/j.foodres.2025.115875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/17/2025] [Accepted: 01/29/2025] [Indexed: 02/24/2025]
Abstract
This study investigated the product characteristics and prebiotic properties of lactose-N-neotetraose (LNnT) after the Maillard reaction with α-lactalbumin (α-LA). LNnT and α-LA were reacted in a simulated system at 60 °C and 79 % relative humidity for 96 h to create Maillard reaction products (MRPs). During the reaction, the absorbances at 294 nm and 420 nm and the fluorescence intensity of α-LA-LNnT increased, whereas the free amino groups and free sugar content decreased. Matrix-assisted laser desorption/ionization time-of-flight/time-of-flight (MALDI-TOF/TOF) analysis revealed that each α-LA was attached with eight LNnT molecules. After the reaction, the far-ultraviolet circular dichroism (CD) and fluorescence spectra showed that the random coil content of α-LA-LNnT significantly increased (p < 0.05), with the side chains of α-LA's tertiary structure affected. In terms of prebiotic properties, the bacterial density of Bifidobacterium decreased from 0.63 to 0.36, and the short-chain fatty acids production decreased from 12.60 mM to 5.38 mM using the in vitro fermentation medium. In summary, various MRPs were formed by the reaction between LNnT and α-LA at different heating stages, and the Maillard reaction reduced the prebiotic properties of LNnT. The findings provide insights into the impact of the Maillard reaction on the prebiotic properties of human milk oligosaccharides and direct our attention to the interactions of nutrients in infant formula.
Collapse
Affiliation(s)
- Simiao Liu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Meixi Liu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Yuzhuo Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Xin Liu
- Department of Nutrition and Health, China Agricultural University, Beijing 100091, China.
| | - Fazheng Ren
- Department of Nutrition and Health, China Agricultural University, Beijing 100091, China; Beijing Laboratory of Food Quality and Safety, Beijing Municipal Education Commission, Beijing 100091, China; Food Laboratory of Zhongyuan, Luohe 462300, Henan, China.
| | - Hao Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Department of Nutrition and Health, China Agricultural University, Beijing 100091, China; Beijing Laboratory of Food Quality and Safety, Beijing Municipal Education Commission, Beijing 100091, China; Food Laboratory of Zhongyuan, Luohe 462300, Henan, China.
| |
Collapse
|
5
|
Weng WC, Liao HE, Chang CH, Hung SC, Du K, Tu Z, Lin CH, Ni CK. Unusual free trisaccharides in caprine colostrum discovered by logically derived sequence tandem mass spectrometry. Sci Rep 2025; 15:1586. [PMID: 39794344 PMCID: PMC11724002 DOI: 10.1038/s41598-024-81561-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/27/2024] [Indexed: 01/13/2025] Open
Abstract
Free oligosaccharides in human milk have many biological functions for infant health. The reducing end of most human milk oligosaccharides is lactose, and caprine milk was reported to contain oligosaccharides structurally similar to those present in human milk. The structures of oligosaccharides were traditionally determined using nuclear magnetic resonance spectroscopy or enzyme digestion followed by various detection methods, e.g., liquid. Mass spectrometry has much higher sensitivity than nuclear magnetic resonance spectroscopy and enzyme digestion. However, conventional mass spectrometry methods only determine part of the structures of oligosaccharides, i.e., compositions and linkage positions. In this study, we used the latest developed mass spectrometry method, namely logically derived sequence tandem mass spectrometry, to determine the complete structures (i.e., composition, linkage positions, anomericities, and stereoisomers) of free neutral trisaccharides in caprine colostrum and mature milk. The high sensitivity of mass spectrometry enables us to discover oligosaccharides of low abundance. Isomers of (Hex)2HexNAc, (Hex)3, and (Hex)2Fuc which have not been reported before were identified. Many of them do not have lactose at the reducing end. Instead, the reducing end is either Glcβ-(1-4)-Glc or Glcβ-(1-4)-GlcNAc. These unusual oligosaccharides are higher in concentration and more structurally diverse in caprine colostrum than that in caprine mature milk and human milk. The structural diversity indicates more complicated biosynthetic pathways of caprine milk compared to that of human milk.
Collapse
Affiliation(s)
- Wei-Chien Weng
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106216, Taiwan
- Molecular Science and Technology, International Graduate Program, Department of Chemistry, Academia Sinica, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Hung-En Liao
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106216, Taiwan
- Department of Appe of Figlied Chemistry, National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan
| | - Cheng-Hsiu Chang
- Genomics Research Center, Academia Sinica, Taipei, 115201, Taiwan
| | - Shang-Cheng Hung
- Genomics Research Center, Academia Sinica, Taipei, 115201, Taiwan
| | - Kai Du
- Institute of Biological Chemistry, Academia Sinica, Taipei, 115201, Taiwan
| | - Zhijay Tu
- Institute of Biological Chemistry, Academia Sinica, Taipei, 115201, Taiwan
| | - Chun-Hung Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei, 115201, Taiwan
- Department of Chemistry, Institute of Biochemical Sciences, National Taiwan University, Taipei, 106319, Taiwan
| | - Chi-Kung Ni
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106216, Taiwan.
| |
Collapse
|
6
|
Duman H, Karav S. Fiber and the gut microbiome and its impact on inflammation. NUTRITION IN THE CONTROL OF INFLAMMATION 2025:51-76. [DOI: 10.1016/b978-0-443-18979-1.00004-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
7
|
Duman H, Bechelany M, Karav S. Human Milk Oligosaccharides: Decoding Their Structural Variability, Health Benefits, and the Evolution of Infant Nutrition. Nutrients 2024; 17:118. [PMID: 39796552 PMCID: PMC11723173 DOI: 10.3390/nu17010118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/24/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Human milk oligosaccharides (HMOs), the third most abundant solid component in human milk, vary significantly among women due to factors such as secretor status, race, geography, season, maternal nutrition and weight, gestational age, and delivery method. In recent studies, HMOs have been shown to have a variety of functional roles in the development of infants. Because HMOs are not digested by infants, they act as metabolic substrates for certain bacteria, helping to establish the infant's gut microbiota. By encouraging the growth of advantageous intestinal bacteria, these sugars function as prebiotics and produce short-chain fatty acids (SCFAs), which are essential for gut health. HMOs can also specifically reduce harmful microbes and viruses binding to the gut epithelium, preventing illness. HMO addition to infant formula is safe and promotes healthy development, infection prevention, and microbiota. Current infant formulas frequently contain oligosaccharides (OSs) that differ structurally from those found in human milk, making it unlikely that they would reproduce the unique effects of HMOs. However, there is a growing trend in producing OSs resembling HMOs, but limited data make it unclear whether HMOs offer additional therapeutic benefits compared to non-human OSs. Better knowledge of how the human mammary gland synthesizes HMOs could direct the development of technologies that yield a broad variety of complex HMOs with OS compositions that closely mimic human milk. This review explores HMOs' complex nature and vital role in infant health, examining maternal variation in HMO composition and its contributing factors. It highlights recent technological advances enabling large-scale studies on HMO composition and its effects on infant health. Furthermore, HMOs' multifunctional roles in biological processes such as infection prevention, brain development, and gut microbiota and immune response regulation are investigated. The structural distinctions between HMOs and other mammalian OSs in infant formulas are discussed, with a focus on the trend toward producing more precise replicas of HMOs found in human milk.
Collapse
Affiliation(s)
- Hatice Duman
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Türkiye;
| | - Mikhael Bechelany
- Institut Européen des Membranes (IEM), UMR 5635, University Montpellier, ENSCM, CNRS, F-34095 Montpellier, France
- Functional Materials Group, Gulf University for Science and Technology (GUST), Masjid Al Aqsa Street, Mubarak Al-Abdullah 32093, Kuwait
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Türkiye;
| |
Collapse
|
8
|
Kesim B, Tezcan Ülger S, Aslan G, Üstün Y, Avcı AT, Küçük MÖ. Effects of Sequential Antimicrobial Phases on Root Canal Microbiome Dynamics in Two-Visit Treatment of Primary Apical Periodontitis: A Longitudinal Experimental Study. Life (Basel) 2024; 14:1696. [PMID: 39768402 PMCID: PMC11680069 DOI: 10.3390/life14121696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Effective management of primary apical periodontitis depends on understanding the dynamic interactions within the root canal microbiome. This study aimed to investigate the effect of sequential antimicrobial phases on the root canal microbiome during a two-visit treatment approach, with a focus on calcium hydroxide medication. METHODS Samples were collected from three teeth across four treatment phases: initial infection (S1), after chemomechanical preparation (S2), after intracanal medication (S3), and after a final flush (S4). DNA was extracted, and the V3-V4 regions of the 16S rRNA gene were sequenced using Illumina MiSeq. Sequencing data were analyzed with QIIME 2, and differentially abundant taxa were identified using linear discriminant analysis effect size (LEfSe). RESULTS While microbial community composition did not differ significantly between phases, the Firmicutes/Bacteroidetes ratio decreased after the antimicrobial stages. LEfSe analysis revealed higher abundances of Lactobacillales, Arthrobacter, and Veillonella in the untreated (CMP) group. Bifidobacterium longum was relatively more abundant in the intracanal medication (ICM) phase, and Dorea formicigenerans was more abundant in the final-flush (FF) phase. CONCLUSIONS Although calcium hydroxide treatment did not induce statistically significant changes in overall root canal microbial composition, trends such as a reduction in the Firmicutes/Bacteroidetes ratio and a relative increase in Bifidobacterium longum numbers suggest potential ecological shifts. The observed relative increase in Bifidobacterium longum numbers may represent a hypothesis-driven observation reflecting indirect ecological effects rather than direct pH modulation. While visual patterns (e.g., PCA clustering) were observed, they lacked statistical support. Further studies with larger sample sizes are needed to validate these observations and assess the potential role of beneficial bacteria in root canal treatments.
Collapse
Affiliation(s)
- Bertan Kesim
- Department of Endodontics, Faculty of Dentistry, Nuh Naci Yazgan University, Kayseri 38170, Turkey
| | - Seda Tezcan Ülger
- Department of Medical Microbiology, Faculty of Medicine, Mersin University, Mersin 33343, Turkey
| | - Gönül Aslan
- Department of Medical Microbiology, Faculty of Medicine, Mersin University, Mersin 33343, Turkey
| | - Yakup Üstün
- Department of Endodontics, Faculty of Dentistry, Erciyes University, Kayseri 38039, Turkey
| | - Ayşe Tuğba Avcı
- Department of Endodontics, Faculty of Dentistry, Erciyes University, Kayseri 38039, Turkey
| | - Mustafa Öner Küçük
- Department of Microbiology, Faculty of Dentistry, Nuh Naci Yazgan University, Kayseri 38170, Turkey
| |
Collapse
|
9
|
Shi Y, Yin R, Pang J, Chen Y, Li Z, Su S, Wen Y. Impact of complementary feeding on infant gut microbiome, metabolites and early development. Food Funct 2024; 15:10663-10678. [PMID: 39354871 DOI: 10.1039/d4fo03948c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Introducing complementary foods is critical for promoting infant health and development. During the weaning period, the dietary patterns provide essential nutrients and facilitate the development of a diverse gut microbiome, which plays significant roles in the regulation of immune, metabolic, and neurological functions. This study enrolled 200 families to assess the impact of complementary feeding on infant growth and health outcomes. Data included detailed records of feeding practices, infant growth measurements, health assessments, and fecal samples and breast milk collected between weeks 12 and 32 postpartum. The gut microbiome was analyzed using 16S rRNA sequencing, while metabolites such as human milk oligosaccharides (HMOs), monosaccharides, and short-chain fatty acids (SCFAs) were measured using chromatography-mass spectrometry. Results revealed a high prevalence of breastfeeding, with complementary food introduced at around 16 weeks. Significant alterations in the infant gut microbiome were observed, particularly in the genera Lactobacillus, Akkermansia, and Staphylococcus. Additionally, the levels of HMOs, monosaccharides, and SCFAs were found to be influenced by the introduction of complementary foods. Significant correlations emerged between complementary feeding practices, gut microbiome diversity, specific bacterial genera (e.g., Streptococcus, Lactobacillus, Bifidobacterium, and Clostridioides), and key metabolites (such as lacto-N-tetraose, lacto-N-neotetraose, mannose, and butyric acid). This study offers valuable insights into the complex interactions between complementary feeding, gut microbiome development, and metabolite profiles during early infant growth. Future research with larger cohorts and targeted dietary interventions is recommended to further elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Yudong Shi
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy Group Co Ltd, China.
| | - Ran Yin
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy Group Co Ltd, China.
| | - Jinzhu Pang
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy Group Co Ltd, China.
| | - Yun Chen
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy Group Co Ltd, China.
| | - Zhouyong Li
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy Group Co Ltd, China.
| | - Shengpeng Su
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy Group Co Ltd, China.
| | - Yongping Wen
- Global R&D Innovation Center, Inner Mongolia Mengniu Dairy Group Co Ltd, China.
| |
Collapse
|
10
|
Mingat S(X, Ehara T, Nakamura H, Miyaji K. Comparative Study of Prebiotics for Infants Using a Fecal Culture System: Insights into Responders and Non-Responders. Nutrients 2024; 16:3347. [PMID: 39408314 PMCID: PMC11478422 DOI: 10.3390/nu16193347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND The gut microbiota of breast-fed infants is dominated by infant-type human-residential bifidobacteria (HRB) that contribute to infant health; thus, it is crucial to develop infant formulas that promote the establishment of a gut microbiota enriched with infant-type HRB, closely resembling that of breastfed infants. METHODS We compared various non-digestible prebiotic oligosaccharides and their combinations using a fecal culture system to explore which candidates could promote the growth of all infant-type HRB and rarely yield non-responders. The analysis included lactulose (LAC), raffinose (RAF), galactooligosaccharides (GOS), and short- and long-chain fructooligosaccharides. Fecal samples were collected from seven infants aged 1.5-10.2 months and cultured with each oligosaccharide individually or their combinations. RESULTS No single oligosaccharide effectively promoted the growth of all infant-type HRB, although GOS promoted the growth of HRB other than Bifidobacterium longum subsp. longum. Only the LAC/RAF/GOS group evenly and effectively promoted the growth of all infant-type HRB. Accordingly, acetate production was higher in fecal cultures supplemented with GOS or LAC/RAF/GOS than in the other cultures, suggesting that it is a superior combination for all infant-type HRB and rarely yields non-responders. CONCLUSIONS This study can aid in developing infant formulas that help align the gut microbiota of formula-fed infants with that of breastfed infants.
Collapse
Affiliation(s)
- Shijir (Xijier) Mingat
- Health Care & Nutritional Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama 252-8583, Japan; (T.E.); (H.N.); (K.M.)
| | | | | | | |
Collapse
|
11
|
Falsaperla R, Sortino V, Gambilonghi F, Vitaliti G, Striano P. Human Milk Oligosaccharides and Their Pivotal Role in Gut-Brain Axis Modulation and Neurologic Development: A Narrative Review to Decipher the Multifaceted Interplay. Nutrients 2024; 16:3009. [PMID: 39275324 PMCID: PMC11397282 DOI: 10.3390/nu16173009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/01/2024] [Accepted: 09/04/2024] [Indexed: 09/16/2024] Open
Abstract
BACKGROUND Human milk oligosaccharides (HMOs), which are unique bioactive components in human milk, are increasingly recognized for their multifaceted roles in infant health. A deeper understanding of the nexus between HMOs and the gut-brain axis can revolutionize neonatal nutrition and neurodevelopmental strategies. METHODS We performed a narrative review using PubMed, Embase, and Google Scholar to source relevant articles. The focus was on studies detailing the influence of HMOs on the gut and brain systems, especially in neonates. Articles were subsequently synthesized based on their exploration into the effects and mechanisms of HMOs on these interconnected systems. RESULTS HMOs significantly influence the neonatal gut-brain axis. Specific concentrations of HMO, measured 1 and 6 months after birth, would seem to agree with this hypothesis. HMOs are shown to influence gut microbiota composition and enhance neurotransmitter production, which are crucial for brain development. For instance, 2'-fucosyllactose has been demonstrated to support cognitive development by fostering beneficial gut bacteria that produce essential short-chain fatty acids. CONCLUSIONS HMOs serve as crucial modulators of the neonatal gut-brain axis, underscoring their importance in infant nutrition and neurodevelopment. Their dual role in shaping the infant gut while influencing brain function presents them as potential game-changers in neonatal health strategies.
Collapse
Affiliation(s)
- Raffaele Falsaperla
- Neonatal Intensive Care Unit and Neonatal Accompaniment Unit, Azienda Ospedaliero-Universitaria Policlinico “Rodolico-San Marco”, San Marco Hospital, University of Catania, 95123 Catania, Italy
- Unit of Pediatrics and Pediatric Emergency, Azienda Ospedaliero-Universitaria Policlinico “Rodolico-San Marco”, San Marco Hospital, University of Catania, 95123 Catania, Italy; (V.S.); (G.V.)
- Department of Medical Science-Pediatrics, University of Ferrara, 44124 Ferrara, Italy
| | - Vincenzo Sortino
- Unit of Pediatrics and Pediatric Emergency, Azienda Ospedaliero-Universitaria Policlinico “Rodolico-San Marco”, San Marco Hospital, University of Catania, 95123 Catania, Italy; (V.S.); (G.V.)
| | - Francesco Gambilonghi
- Postgraduate Training Program in Pediatrics, Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy;
| | - Giovanna Vitaliti
- Unit of Pediatrics and Pediatric Emergency, Azienda Ospedaliero-Universitaria Policlinico “Rodolico-San Marco”, San Marco Hospital, University of Catania, 95123 Catania, Italy; (V.S.); (G.V.)
| | - Pasquale Striano
- IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy;
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16126 Genoa, Italy
| |
Collapse
|
12
|
Lee HY, Nazmul T, Lan J, Oyoshi MK. Maternal influences on offspring food allergy. Immunol Rev 2024; 326:130-150. [PMID: 39275992 PMCID: PMC11867100 DOI: 10.1111/imr.13392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2024]
Abstract
The prevalence of allergies has been globally escalating. While allergies could appear at any age, they often develop in early life. However, the significant knowledge gap in the field is the mechanisms by which allergies affect certain people but not others. Investigating early factors and events in neonatal life that have a lasting impact on determining the susceptibilities of children to develop allergies is a significant area of the investigation as it promotes the understanding of neonatal immune system that mediates tolerance versus allergies. This review focuses on the research over the recent 10 years regarding the potential maternal factors that influence offspring allergies with a view to food allergy, a potentially life-threatening cause of anaphylaxis. The role of breast milk, maternal diet, maternal antibodies, and microbiota that have been suggested as key maternal factors regulating offspring allergies are discussed here. We also suggest future research area to expand our knowledge of maternal-offspring interactions on the pathogenesis of food allergy.
Collapse
Affiliation(s)
- Hwa Yeong Lee
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Tanuza Nazmul
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
| | - Jinggang Lan
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
| | - Michiko K. Oyoshi
- Division of Pediatric Allergy, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Charlestown, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Tewari N, Dey P. Navigating commensal dysbiosis: Gastrointestinal host-pathogen interplay orchestrating opportunistic infections. Microbiol Res 2024; 286:127832. [PMID: 39013300 DOI: 10.1016/j.micres.2024.127832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/23/2024] [Accepted: 07/01/2024] [Indexed: 07/18/2024]
Abstract
The gut commensals, which are usually symbiotic or non-harmful bacteria that live in the gastrointestinal tract, have a positive impact on the health of the host. This review, however, specifically discuss distinct conditions where commensals aid in the development of pathogenic opportunistic infections. We discuss that the categorization of gut bacteria as either pathogens or non-pathogens depends on certain circumstances, which are significantly affected by the tissue microenvironment and the dynamic host-microbe interaction. Under favorable circumstances, commensals have the ability to transform into opportunistic pathobionts by undergoing overgrowth. These conditions include changes in the host's physiology, simultaneous infection with other pathogens, effective utilization of nutrients, interactions between different species of bacteria, the formation of protective biofilms, genetic mutations that enhance pathogenicity, acquisition of genes associated with virulence, and the ability to avoid the host's immune response. These processes allow commensals to both initiate infections themselves and aid other pathogens in populating the host. This review highlights the need of having a detailed and sophisticated knowledge of the two-sided nature of gut commensals. Although commensals mostly promote health, they may also become harmful in certain changes in the environment or the body's functioning. This highlights the need of acknowledging the intricate equilibrium in interactions between hosts and microbes, which is crucial for preserving intestinal homeostasis and averting diseases. Finally, we also emphasize the further need of research to better understand and anticipate the behavior of gut commensals in different situations, since they play a crucial and varied role in human health and disease.
Collapse
Affiliation(s)
- Nisha Tewari
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab 147004, India
| | - Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab 147004, India.
| |
Collapse
|
14
|
Du Z, Li Z, Guang C, Zhu Y, Mu W. Recent advances of 3-fucosyllactose in health effects and production. Arch Microbiol 2024; 206:378. [PMID: 39143417 DOI: 10.1007/s00203-024-04104-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 08/16/2024]
Abstract
Human milk oligosaccharides (HMOs) have been recognized as gold standard for infant development. 3-Fucosyllactose (3-FL), being one of the Generally Recognized as Safe HMOs, represents a core trisaccharide within the realm of HMOs; however, it has received comparatively less attention in contrast to extensively studied 2'-fucosyllactose. The objective of this review is to comprehensively summarize the health effects of 3-FL, including its impact on gut microbiota proliferation, antimicrobial effects, immune regulation, antiviral protection, and brain maturation. Additionally, the discussion also covers the commercial application and regulatory approval status of 3-FL. Lastly, an organized presentation of large-scale production methods for 3-FL aims to provide a comprehensive guide that highlights current strategies and challenges in optimization.
Collapse
Affiliation(s)
- Zhihui Du
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China
| | - Zeyu Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China
| | - Cuie Guang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China
| | - Yingying Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China.
| |
Collapse
|
15
|
Mulinge MM, Abisi HK, Kabahweza HM, Okutoyi L, Wamalwa DC, Nduati RW. The Role of Maternal Secretor Status and Human Milk Oligosaccharides on Early Childhood Development: A Systematic Review and Meta-Analysis. Breastfeed Med 2024; 19:409-424. [PMID: 38577928 DOI: 10.1089/bfm.2023.0274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Background: Breast milk is the gold standard of infant nutrition, delivering nutrients and bioactive molecules as needed to support optimal infant growth and cognitive development. Increasing evidence links human milk oligosaccharides (HMOs) to these early childhood development milestones. Aims: To summarize and synthesize the evidence relating to HMOs and infant brain development, physical growth, and cognitive development. In addition, HMO concentrations in secretor and nonsecretor mothers were compared via a meta-analysis. Study Design: A systematic review and meta-analysis were carried out in accordance with the PRISMA statement. This review used three databases (PubMed, Scopus, and Web of Science) and was limited to English-language articles published between 2000 and June 30, 2023. Results: The initial searches yielded 245 articles, 27 of which were included in the systematic review and 12 in the meta-analysis. The meta-analysis revealed a substantial between-study heterogeneity, I2 = 97.3%. The pooled effect was 0.21 (95% CI: -0.41 to 0.83; p = 0.484), indicating that secretors had higher HMO concentrations, although this difference was not statistically significant. At one month of age, 2'FL, 3FL, and 3'SL play an important role in brain maturation and thus play a critical role in cognitive development. Secretors produce higher concentrations of 2'FL and 3'SL, explaining the benefits to infants of secretor mothers. Growth velocity was correlated to fucosylated and sialylated HMO concentrations, with lower concentrations linked to stunting. Conclusions: According to evidence from the systematically reviewed articles, HMOs are essential for a child's early development, but the extent to which they have an impact depends on maternal secretor status.
Collapse
Affiliation(s)
- Martin M Mulinge
- Department of Biochemistry, School of Medicine, University of Nairobi, Nairobi, Kenya
| | - Hellen K Abisi
- Department of Biochemistry, School of Medicine, University of Nairobi, Nairobi, Kenya
| | - Hellen M Kabahweza
- Department of Pediatric Hematology & Oncology, Joint Clinical Research Centre, Kampala, Uganda
| | - Lydia Okutoyi
- Department of Health Care Quality, Kenyatta National Hospital, Nairobi, Kenya
| | - Dalton C Wamalwa
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Ruth W Nduati
- Department of Paediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| |
Collapse
|
16
|
Nguan HS, Chen JL, Ni CK. Collision-Induced Dissociation of Fucose and Identification of Anomericity. J Phys Chem A 2024; 128:3812-3820. [PMID: 38690855 PMCID: PMC11103703 DOI: 10.1021/acs.jpca.4c00640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/15/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024]
Abstract
Structural determination of carbohydrates using mass spectrometry remains challenging, particularly, the differentiation of anomeric configurations. In this work, we studied the collision-induced dissociation (CID) mechanisms of sodiated α- and β-l-fucose using an experimental method and quantum chemistry calculations. The calculations show that α-l-fucose is more likely to undergo dehydration due to the fact that O1 and O2 are on the same side of the sugar ring. In contrast, β-l-fucose is more prone to the ring-opening reaction because more OH groups are on the same side of the sugar ring as O1. These differences suggest a higher preference for the dehydration reaction in sodiated α-l-fucose but a lower preference for ring-opening compared to that of β-l-fucose. The calculation results, which are used to assign the CID mass spectra of α- and β-l-fucose separated by high-performance liquid chromatography, are supported by the fucose produced from the CID of disaccharides Fuc-β-(1 → 3)-GlcNAc and Fuc-α-(1 → 4)-GlcNAc. This study demonstrates that the correlation of cis- and trans-configurations of O1 and O2 to the relative branching ratios of dehydration and cross-ring dissociation in CID, observed in aldohexose and ketohexose in the pyranose form, can be extended to deoxyhexoses for anomericity determination.
Collapse
Affiliation(s)
- Hock-Seng Nguan
- Institute
of Atomic and Molecular Sciences, Academia
Sinica, P.O. Box 23-166, Taipei 10617, Taiwan
| | - Jien-Lian Chen
- Institute
of Atomic and Molecular Sciences, Academia
Sinica, P.O. Box 23-166, Taipei 10617, Taiwan
| | - Chi-Kung Ni
- Institute
of Atomic and Molecular Sciences, Academia
Sinica, P.O. Box 23-166, Taipei 10617, Taiwan
- Department
of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
| |
Collapse
|
17
|
Thorman AW, Morrow AL, Groeneveld A, Nauta A, Newburg DS. Validation of collection and anaerobic fermentation techniques for measuring prebiotic impact on gut microbiota. Pharmacol Res 2024; 203:107169. [PMID: 38583688 DOI: 10.1016/j.phrs.2024.107169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/25/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Defining the ability of prebiotic dietary carbohydrates to influence the composition and metabolism of the gut microbiota is central to defining their health impact in diverse individuals. Many clinical trials are using indirect methods. This study aimed to validate collection and fermentation methods enabling their use in the context of clinical studies. METHODS AND RESULTS Parameters tested included stool sample acquisition, storage, and growth conditions. Stool from 3 infants and 3 adults was collected and stored under varying conditions. Samples were cultured anaerobically for two days in the presence of prebiotics, whereupon optical density and pH were measured across time. Whole genome shotgun sequencing and NMR metabolomics were performed. Neither the type of collection vial (standard vial and two different BD anaerobic collection vials) nor cryopreservation (-80 °C or 4 °C) significantly influenced either microbial composition at 16 h of anaerobic culture or the principal components of the metabolome at 8 or 16 h. Metagenomic differences were driven primarily by subject, while metabolomic differences were driven by fermentation sugar (2'-fucosyllactose or dextrose). CONCLUSIONS These data identified a feasible and valid approach for prebiotic fermentation analysis of individual samples in large clinical studies: collection of stool microbiota using standard vials; cryopreservation prior to testing; and collecting fermentation read-out at 8 and 16 hr. Thus, fermentation analysis can be a valid technique for testing the effects of prebiotics on human fecal microbiota.
Collapse
Affiliation(s)
- Alexander W Thorman
- University of Cincinnati College of Medicine, Department of Environmental and Public Health Sciences, Kettering Labs, 160 Panzeca Way, Cincinnati, OH 45267, United States.
| | - Ardythe L Morrow
- University of Cincinnati College of Medicine, Department of Environmental and Public Health Sciences, Kettering Labs, 160 Panzeca Way, Cincinnati, OH 45267, United States.
| | - Andre Groeneveld
- FrieslandCampina, Bronland 20, Wageningen 6708 WH, the Netherlands.
| | - Arjen Nauta
- FrieslandCampina, Bronland 20, Wageningen 6708 WH, the Netherlands.
| | - David S Newburg
- University of Cincinnati College of Medicine, Department of Environmental and Public Health Sciences, Kettering Labs, 160 Panzeca Way, Cincinnati, OH 45267, United States.
| |
Collapse
|
18
|
Zhou Y, Liu X, Chen H, Zhao J, Zhang H, Chen W, Yang B. Isolation and Characterisation of Streptococcus spp. with Human Milk Oligosaccharides Utilization Capacity from Human Milk. Foods 2024; 13:1291. [PMID: 38731662 PMCID: PMC11083076 DOI: 10.3390/foods13091291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024] Open
Abstract
Human milk oligosaccharides (HMO) that promote the growth of beneficial gut microbes in infants are abundant in human milk. Streptococcus, one of the dominant genera in human milk microbiota, is also highly prevalent in the infant gut microbiota, possibly due to its adeptness at utilizing HMOs. While previous studies have mainly focused on HMO interactions with gut bacteria like Bifidobacterium and Bacteroides spp., the interaction with Streptococcus spp. has not been fully explored. In this study, Streptococcus spp. was isolated from human milk and identified to exhibit extensive capabilities in utilizing HMOs. Their consumption rates of 2'-fucosyllactose (2'-FL), 6'-sialyllactose (6'-SL), and lacto-N-tetraose (LNT) closely matched those of Bifidobacterium longum subsp. infantis ATCC 15697. Furthermore, we assessed the safety-related genes in the genomes of the Streptococcus species capable of utilizing HMOs, revealing potential virulence and resistance genes. In addition, no haemolytic activity was observed. These findings expand the knowledge of metabolic interactions and networks within the microbiota of human milk and the early life human gut.
Collapse
Affiliation(s)
- Ye Zhou
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.Z.); (X.L.); (H.C.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xiaoming Liu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.Z.); (X.L.); (H.C.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Haiqin Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.Z.); (X.L.); (H.C.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.Z.); (X.L.); (H.C.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.Z.); (X.L.); (H.C.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.Z.); (X.L.); (H.C.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Bo Yang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.Z.); (X.L.); (H.C.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
19
|
Chen Y, Chen Z, Zhu Y, Wen Y, Zhao C, Mu W. Recent Progress in Human Milk Oligosaccharides and Its Antiviral Efficacy. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:7607-7617. [PMID: 38563422 DOI: 10.1021/acs.jafc.3c09460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Gastrointestinal (GI)-associated viruses, including rotavirus (RV), norovirus (NV), and enterovirus, usually invade host cells, transmit, and mutate their genetic information, resulting in influenza-like symptoms, acute gastroenteritis, encephalitis, or even death. The unique structures of human milk oligosaccharides (HMOs) enable them to shape the gut microbial diversity and endogenous immune system of human infants. Growing evidence suggests that HMOs can enhance host resistance to GI-associated viruses but without a systematic summary to review the mechanism. The present review examines the lactose- and neutral-core HMOs and their antiviral effects in the host. The potential negative impacts of enterovirus 71 (EV-A71) and other GI viruses on children are extensive and include neurological sequelae, neurodevelopmental retardation, and cognitive decline. However, the differences in the binding affinity of HMOs for GI viruses are vast. Hence, elucidating the mechanisms and positive effects of HMOs against different viruses may facilitate the development of novel HMO derived oligosaccharides.
Collapse
Affiliation(s)
- Yihan Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Zhengxin Chen
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yingying Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| | - Yuxi Wen
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Department of Analytical and Food Chemistry, Faculty of Sciences, Universidade de Vigo, 32004 Ourense Spain
| | - Chao Zhao
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
20
|
Zhu Y, Chen R, Wang H, Chen Y, Huang Z, Du Z, Meng J, Zhou J, Mu W. De Novo Biosynthesis of Difucosyllactose by Artificial Pathway Construction and α1,3/4-Fucosyltransferase Rational Design in Escherichia coli. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 38598361 DOI: 10.1021/acs.jafc.4c01691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Difucosyllactose (DFL) is a significant and plentiful oligosaccharide found in human breast milk. In this study, an artificial metabolic pathway of DFL was designed, focusing on the de novo biosynthesis of GDP-fucose from only glycerol. This was achieved by engineering Escherichia coli to endogenously overexpress genes manB, manC, gmd, and wcaG and heterologously overexpress a pair of fucosyltransferases to produce DFL from lactose. The introduction of α-1,2-fucosyltransferase from Helicobacter pylori (FucT2) along with α-1,3/4-fucosyltransferase (HP3/4FT) addressed rate-limiting challenges in enzymatic catalysis and allowed for highly efficient conversion of lactose into DFL. Based on these results, molecular modification of HP3/4FT was performed based on computer-assisted screening and structure-based rational design. The best-performing mutant, MH5, containing a combination of five mutated sites (F49K/Y131D/Y197N/E338D/R369A) of HP3/4FT was obtained. The best strain BLC09-58 harboring MH5 yielded 45.81 g/L of extracellular DFL in 5-L fed-batch cultures, which was the highest titer reported to date.
Collapse
Affiliation(s)
- Yingying Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Roulin Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Hao Wang
- Bloomage Biotechnology Corp., Ltd., Jinan, Shandong 250010, People's Republic of China
| | - Yihan Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Zhaolin Huang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Zhihui Du
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Jiawei Meng
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Jingwen Zhou
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, People's Republic of China
| |
Collapse
|
21
|
Ma L, Zhu Y, Zhu La ALT, Lourenco JM, Callaway TR, Bu D. Schizochytrium sp. and lactoferrin supplementation alleviates Escherichia coli K99-induced diarrhea in preweaning dairy calves. J Dairy Sci 2024; 107:1603-1619. [PMID: 37769949 DOI: 10.3168/jds.2023-23466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 09/04/2023] [Indexed: 10/03/2023]
Abstract
Calf diarrhea, a common disease mainly induced by Escherichia coli infection, is one of the main reasons for nonpredator losses. Hence, an effective nonantibacterial approach to prevent calf diarrhea has become an emerging requirement. This study evaluated the microalgae Schizochytrium sp. (SZ) and lactoferrin (LF) as a nutrient intervention approach against E. coli O101:K99-induced preweaning calve diarrhea. Fifty 1-d-old male Holstein calves were randomly divided into 5 groups (n = 10): (1) control, (2) blank (no supplement or challenge), (3) 1 g/d LF, (4) 20 g/d SZ, or (5) 1 g/d LF plus 20 g/d SZ (LFSZ). The experimental period lasted 14 d. On the morning of d 7, calves were challenged with 1 × 1011 cfu of E. coli O101:K99, and rectum feces were collected on 3, 12, 24, and 168 h postchallenge for the control, LF, SZ, and LFSZ groups. The rectal feces of the blank group were collected on d 14. Data were analyzed using the mixed procedure of SAS (version 9.4; SAS Institute Inc.). The E. coli K99 challenge decreased the average daily gain (ADG) and increased feed-to-gain ratio (F:G) and diarrhea frequency (control vs. blank). Compared with the control group, the LFSZ group had a higher ADG and lower F:G, and the LFSZ and SZ groups had lower diarrhea frequency compared with the control group. In addition, the LFSZ and SZ groups have no differences in diarrhea frequency compared with the blank group. Compared with the control group, the blank group had lower serum nitric oxide (NO), endothelin-1, d-lactic acid (D-LA), and lipopolysaccharide (LPS) concentrations, as well as serum IgG, IL-1β, IL-6, IL-10, and TNF-α levels on d 7 and 14. On d 7, compared with the control group, all treatment groups had lower serum NO level, the SZ group had a lower serum D-LA concentration, and the LF and LFSZ groups had lower serum LPS concentration. On d 14, compared with the control group, the fecal microbiota of the blank group had lower Shannon, Simpson, Chao1, and ACE indexes, the LFSZ group had lower Shannon and Simpson indexes, the SZ and LFSZ groups had a higher Chao1 index, and all treatment groups had a higher ACE index. In fecal microbiota, Bifidobacterium and Actinobacteria were negatively associated with IL-10 and d-lactate, while Akkermansia was negatively associated with endothelin-1 and positively correlated with LPS, fecal scores, and d-lactate levels. Our results indicated that LF and SZ supplements could alleviate E. coli O101:K99-induced calf diarrhea individually or in combination. Supplementing 1 g/d LF and 20 g/d SZ could be a potential nutrient intervention approach to prevent bacterial diarrhea in calves.
Collapse
Affiliation(s)
- Lu Ma
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yingkun Zhu
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; School of Agriculture & Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - A La Teng Zhu La
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - J M Lourenco
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602
| | - T R Callaway
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602
| | - Dengpan Bu
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; CAAS-ICRAF Joint Lab on Agroforestry and Sustainable Animal Husbandry, World Agroforestry Centre, East and Central Asia, Beijing 100193, China.
| |
Collapse
|
22
|
Kuntz S, Kunz C, Borsch C, Hill D, Morrin S, Buck R, Rudloff S. Influence of microbially fermented 2´-fucosyllactose on neuronal-like cell activity in an in vitro co-culture system. Front Nutr 2024; 11:1351433. [PMID: 38389793 PMCID: PMC10881714 DOI: 10.3389/fnut.2024.1351433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 01/18/2024] [Indexed: 02/24/2024] Open
Abstract
Scope 2´-Fucosyllactose (2´-FL), the most abundant oligosaccharide in human milk, plays an important role in numerous biological functions, including improved learning. It is not clear, however, whether 2´-FL or a cleavage product could influence neuronal cell activity. Thus, we investigated the effects of 2´-FL, its monosaccharide fucose (Fuc), and microbial fermented 2´-FL and Fuc on the parameters of neuronal cell activity in an intestinal-neuronal transwell co-culture system in vitro. Methods Native 13C-labeled 2´-FL and 13C-Fuc or their metabolites, fermented with Bifidobacterium (B.) longum ssp. infantis and B. breve, which were taken from the lag-, log- and stationary (stat-) growth phases of batch cultures, were applied to the apical compartment of the co-culture system with Caco-2 cells representing the intestinal layer and all-trans-retinoic acid-differentiated SH-SY5Y (SH-SY5YATRA) cells mimicking neuronal-like cells. After 3 h of incubation, the culture medium in the basal compartment was monitored for 13C enrichment by using elemental analysis isotope-ratio mass spectrometry (EA-IRMS) and effects on cell viability, plasma, and mitochondrial membrane potential. The neurotransmitter activation (BDNF, GABA, choline, and glutamate) of SH-SY5YATRA cells was also determined. Furthermore, these effects were also measured by the direct application of 13C-2´-FL and 13C-Fuc to SH-SY5YATRA cells. Results While no effects on neuronal-like cell activities were observed after intact 2´-FL or Fuc was incubated with SH-SY5YATRA cells, supernatants from the stat-growth phase of 2´-FL, fermented by B. longum ssp. infantis alone and together with B. breve, significantly induced BDNF release from SH-SY5YATRA cells. No such effects were found for 2´-FL, Fuc, or their fermentation products from B. breve. The BDNF release occurred from an enhanced vesicular release, which was confirmed by the use of the Ca2+-channel blocker verapamil. Concomitant with this event, 13C enrichment was also observed in the basal compartment when supernatants from the stat-growth phase of fermentation by B. longum ssp. infantis alone or together with B. breve were used. Conclusion The results obtained in this study suggest that microbial products of 2´-FL rather than the oligosaccharide itself may influence neuronal cell activities.
Collapse
Affiliation(s)
- Sabine Kuntz
- Department of Nutritional Science, Justus Liebig University Giessen, Giessen, Germany
| | - Clemens Kunz
- Department of Nutritional Science, Justus Liebig University Giessen, Giessen, Germany
| | - Christian Borsch
- Department of Nutritional Science, Justus Liebig University Giessen, Giessen, Germany
| | - David Hill
- Abbott, Nutrition Division, Columbus, OH, United States
| | - Sinéad Morrin
- Abbott, Nutrition Division, Columbus, OH, United States
| | - Rachael Buck
- Abbott, Nutrition Division, Columbus, OH, United States
| | - Silvia Rudloff
- Department of Nutritional Science, Justus Liebig University Giessen, Giessen, Germany
- Department of Pediatrics, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
23
|
Zhang Y, Ye Y, Guo J, Wang M, Li X, Ren Y, Zhu W, Yu K. Effects of 2'-fucosyllactose on the composition and metabolic activity of intestinal microbiota from piglets after in vitro fermentation. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:1553-1563. [PMID: 37815100 DOI: 10.1002/jsfa.13037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 09/27/2023] [Accepted: 10/10/2023] [Indexed: 10/11/2023]
Abstract
BACKGROUND As indigestible carbohydrates, milk oligosaccharides possess various benefits for newborns, mainly through intestinal microbiota, among which 2'-fucosyllactose (2'-FL) is the most predominant milk oligosaccharide. However, knowledge about the fermentative characteristics of 2'-FL in the gut remains limited, especially in the small intestine. The aim of this study is to explore the differential fermentability of 2'-FL by the small and large intestinal microbiota of piglets using fructo-oligosaccharide (FOS) and lactose as controls in an in vitro batch fermentation experiment. During fermentation, microbial composition was characterized along with gas production and short-chain fatty acid production. RESULTS 2'-Fucosyllactose showed differential fermentability in jejunal and colonic fermentation. Compared with the colon, 2'-FL produced less gas in the jejunum than in the FOS and lactose groups (P < 0.05). Meanwhile, 2'-FL exhibited a different influence on the microbial composition and metabolism in the jejunum and colon compared with FOS and lactose. In the jejunum, compared with the FOS and lactose groups, the 2'-FL group showed a higher abundance of Bacteroides, Prevotella, and Blautia, but a lower abundance of Streptococcus and Lactobacillus (P < 0.05), with a higher level of propionate and a lower level of lactate during fermentation (P < 0.05). In the colon, compared with the FOS and lactose groups, 2'-FL increased the abundance of Blautia, Faecalibacterium, and Lachnospiraceae FCS020, but decreased the abundance of Prevotella_9, Succinivibrio, and Megasphaera (P < 0.05) with an increase in acetate production (P < 0.05). CONCLUSION Overall, the results suggested that the small intestinal microbiota had the potential to ferment milk oligosaccharides. Meanwhile, in comparison with FOS and lactose, 2'-FL selectively stimulated the growth of propionate-producing bacteria in the jejunum and acetate-producing bacteria in the colon. These results demonstrated the differences in fermentation properties of 2'-FL by small and large intestinal microbiota and provided new evidence for the application of 2'-FL in optimizing gut microbiota. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yanan Zhang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China
| | - Yanxin Ye
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Jiaqing Guo
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Mengting Wang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Xuan Li
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Yuting Ren
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Kaifan Yu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
24
|
Wu H, Mu C, Xu L, Yu K, Shen L, Zhu W. Host-microbiota interaction in intestinal stem cell homeostasis. Gut Microbes 2024; 16:2353399. [PMID: 38757687 PMCID: PMC11110705 DOI: 10.1080/19490976.2024.2353399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/06/2024] [Indexed: 05/18/2024] Open
Abstract
Intestinal stem cells (ISCs) play a pivotal role in gut physiology by governing intestinal epithelium renewal through the precise regulation of proliferation and differentiation. The gut microbiota interacts closely with the epithelium through myriad of actions, including immune and metabolic interactions, which translate into tight connections between microbial activity and ISC function. Given the diverse functions of the gut microbiota in affecting the metabolism of macronutrients and micronutrients, dietary nutrients exert pronounced effects on host-microbiota interactions and, consequently, the ISC fate. Therefore, understanding the intricate host-microbiota interaction in regulating ISC homeostasis is imperative for improving gut health. Here, we review recent advances in understanding host-microbiota immune and metabolic interactions that shape ISC function, such as the role of pattern-recognition receptors and microbial metabolites, including lactate and indole metabolites. Additionally, the diverse regulatory effects of the microbiota on dietary nutrients, including proteins, carbohydrates, vitamins, and minerals (e.g. iron and zinc), are thoroughly explored in relation to their impact on ISCs. Thus, we highlight the multifaceted mechanisms governing host-microbiota interactions in ISC homeostasis. Insights gained from this review provide strategies for the development of dietary or microbiota-based interventions to foster gut health.
Collapse
Affiliation(s)
- Haiqin Wu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Chunlong Mu
- Food Informatics, AgResearch, Te Ohu Rangahau Kai, Palmerston North, New Zealand
| | - Laipeng Xu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Kaifan Yu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Le Shen
- Department of Surgery, The University of Chicago, Chicago, IL, USA
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
25
|
Lee YG, Jo HY, Lee DH, Yoon JW, Song YH, Kweon DH, Kim KH, Park YC, Seo JH. De novo biosynthesis of 2'-fucosyllactose by bioengineered Corynebacterium glutamicum. Biotechnol J 2024; 19:e2300461. [PMID: 37968827 DOI: 10.1002/biot.202300461] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/27/2023] [Accepted: 11/12/2023] [Indexed: 11/17/2023]
Abstract
2'-Fucosyllactose (2'-FL) which is well-known human milk oligosaccharide was biotechnologically synthesized using engineered Corynebacterium glutamicum, a GRAS microbial workhorse. By construction of the complete de novo pathway for GDP-L-fucose supply and heterologous expression of Escherichia coli lactose permease and Helicobacter pylori α-1,2-fucosyltransferase, bioengineered C. glutamicum BCGW_TL successfully biosynthesized 0.25 g L-1 2'-FL from glucose. The additional genetic perturbations including the expression of a putative 2'-FL exporter and disruption of the chromosomal pfkA gene allowed C. glutamicum BCGW_cTTLEΔP to produce 2.5 g L-1 2'-FL batchwise. Finally, optimized fed-batch cultivation of the BCGW_cTTLEΔP using glucose, fructose, and lactose resulted in 21.5 g L-1 2'-FL production with a productivity of 0.12 g L-1 •h, which were more than 3.3 times higher value relative to the batch culture of the BCGW_TL. Conclusively, it would be a groundwork to adopt C. glutamicum for biotechnological production of other food additives including human milk oligosaccharides.
Collapse
Affiliation(s)
- Ye-Gi Lee
- Department of Bio and Fermentation Convergence Technology and Center for Bioconvergence, Kookmin University, Seoul, South Korea
- Department of Agricultural Biotechnology and Center for Food Bioconvergence, Seoul National University, Seoul, South Korea
| | - Hae-Yong Jo
- Department of Agricultural Biotechnology and Center for Food Bioconvergence, Seoul National University, Seoul, South Korea
| | - Do-Haeng Lee
- Department of Agricultural Biotechnology and Center for Food Bioconvergence, Seoul National University, Seoul, South Korea
| | - Jong-Won Yoon
- Advanced Protein Technologies Corp. Yeongtong-gu, Suwon, Gyeonggi, South Korea
| | - Young-Ha Song
- Advanced Protein Technologies Corp. Yeongtong-gu, Suwon, Gyeonggi, South Korea
| | - Dae-Hyuk Kweon
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, South Korea
| | - Kyoung Heon Kim
- Department of Biotechnology, Graduate School, Korea University, Seoul, South Korea
| | - Yong-Cheol Park
- Department of Bio and Fermentation Convergence Technology and Center for Bioconvergence, Kookmin University, Seoul, South Korea
| | - Jin-Ho Seo
- Department of Agricultural Biotechnology and Center for Food Bioconvergence, Seoul National University, Seoul, South Korea
| |
Collapse
|
26
|
Dey P, Ray Chaudhuri S. The opportunistic nature of gut commensal microbiota. Crit Rev Microbiol 2023; 49:739-763. [PMID: 36256871 DOI: 10.1080/1040841x.2022.2133987] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 07/30/2022] [Accepted: 10/05/2022] [Indexed: 11/03/2022]
Abstract
The abundance of gut commensals has historically been associated with health-promoting effects despite the fact that the definition of good or bad microbiota remains condition-specific. The beneficial or pathogenic nature of microbiota is generally dictated by the dimensions of host-microbiota and microbe-microbe interactions. With the increasing popularity of gut microbiota in human health and disease, emerging evidence suggests opportunistic infections promoted by those gut bacteria that are generally considered beneficial. Therefore, the current review deals with the opportunistic nature of the gut commensals and aims to summarise the concepts behind the occasional commensal-to-pathogenic transformation of the gut microbes. Specifically, relevant clinical and experimental studies have been discussed on the overgrowth and bacteraemia caused by commensals. Three key processes and their underlying mechanisms have been summarised to be responsible for the opportunistic nature of commensals, viz. improved colonisation fitness that is dictated by commensal-pathogen interactions and availability of preferred nutrients; pathoadaptive mutations that can trigger the commensal-to-pathogen transformation; and evasion of host immune response as a survival and proliferation strategy of the microbes. Collectively, this review provides an updated concept summary on the underlying mechanisms of disease causative events driven by gut commensal bacteria.
Collapse
Affiliation(s)
- Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Saumya Ray Chaudhuri
- Council of Scientific and Industrial Research (CSIR), Institute of Microbial Technology, Chandigarh, India
| |
Collapse
|
27
|
Jacobs JP, Lee ML, Rechtman DJ, Sun AK, Autran C, Niklas V. Human milk oligosaccharides modulate the intestinal microbiome of healthy adults. Sci Rep 2023; 13:14308. [PMID: 37652940 PMCID: PMC10471580 DOI: 10.1038/s41598-023-41040-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 08/18/2023] [Indexed: 09/02/2023] Open
Abstract
Human milk contains over 200 distinct oligosaccharides, which are critical to shaping the developing neonatal gut microbiome. To investigate whether a complex mixture of human milk oligosaccharides (HMOs) would similarly modulate the adult gut microbiome, HMO-Concentrate derived from pooled donor breast milk was administered orally to 32 healthy adults for 7 days followed by 21 days of monitoring. Fecal samples were collected for 16S rRNA gene sequencing, shotgun metagenomics, and metabolomics analyses. HMO-Concentrate induced dose-dependent Bifidobacterium expansion, reduced microbial diversity, and altered microbial gene content. Following HMO cessation, a microbial succession occurred with diverse taxonomic changes-including Bacteroides expansion-that persisted through day 28. This was associated with altered microbial gene content, shifts in serum metabolite levels, and increased circulating TGFβ and IL-10. Incubation of cultured adult microbiota with HMO-Concentrate induced dose-dependent compositional shifts that were not recapitulated by individual HMOs or defined mixtures of the 10 most abundant HMOs in HMO-Concentrate at their measured concentrations. These findings support that pooled donor HMOs can exert direct effects on adult gut microbiota and that complex mixtures including low abundance HMOs present in donor milk may be required for maximum effect.Registration: ClinicalTrials.gov NCT05516225.
Collapse
Affiliation(s)
- Jonathan P Jacobs
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA.
- Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA.
| | - Martin L Lee
- Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA
- Prolacta Bioscience, Duarte, CA, USA
| | | | | | | | - Victoria Niklas
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Oak Hill Bio Ltd, Altrincham, Cheshire WA14 2DT, United Kingdom
| |
Collapse
|
28
|
Cho S, Samuel TM, Li T, Howell BR, Baluyot K, Hazlett HC, Elison JT, Zhu H, Hauser J, Sprenger N, Lin W. Interactions between Bifidobacterium and Bacteroides and human milk oligosaccharides and their associations with infant cognition. Front Nutr 2023; 10:1216327. [PMID: 37457984 PMCID: PMC10345227 DOI: 10.3389/fnut.2023.1216327] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
While ample research on independent associations between infant cognition and gut microbiota composition and human milk (HM) oligosaccharides (HMOs) has been reported, studies on how the interactions between gut microbiota and HMOs may yield associations with cognitive development in infancy are lacking. We aimed to determine how HMOs and species of Bacteroides and Bifidobacterium genera interact with each other and their associations with cognitive development in typically developing infants. A total of 105 mother-infant dyads were included in this study. The enrolled infants [2.9-12 months old (8.09 ± 2.48)] were at least predominantly breastfed at 4 months old. A total of 170 HM samples from the mothers and fecal samples of the children were collected longitudinally. Using the Mullen Scales of Early Learning to assess cognition and the scores as the outcomes, linear mixed effects models including both the levels of eight HMOs and relative abundance of Bacteroides and Bifidobacterium species as main associations and their interactions were employed with adjusting covariates; infant sex, delivery mode, maternal education, site, and batch effects of HMOs. Additionally, regression models stratifying infants based on the A-tetrasaccharide (A-tetra) status of the HM they received were also employed to determine if the associations depend on the A-tetra status. With Bacteroides species, we observed significant associations with motor functions, while Bif. catenulatum showed a negative association with visual reception in the detectable A-tetra group both as main effect (value of p = 0.012) and in interaction with LNFP-I (value of p = 0.007). Additionally, 3-FL showed a positive association with gross motor (p = 0.027) and visual reception (p = 0.041). Furthermore, significant associations were observed with the interaction terms mainly in the undetectable A-tetra group. Specifically, we observed negative associations for Bifidobacterium species and LNT [breve (p = 0.011) and longum (p = 0.022)], and positive associations for expressive language with 3'-SL and Bif. bifidum (p = 0.01), 6'-SL and B. fragilis (p = 0.019), and LNFP-I and Bif. kashiwanohense (p = 0.048), respectively. Our findings suggest that gut microbiota and HMOs are both independently and interactively associated with early cognitive development. In particular, the diverse interactions between HMOs and Bacteroides and Bifidobacterium species reveal different candidate pathways through which HMOs, Bifidobacterium and Bacteroides species potentially interact to impact cognitive development in infancy.
Collapse
Affiliation(s)
- Seoyoon Cho
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Tinu M. Samuel
- Nestle Product Technology Center-Nutrition, Société des Produits Nestlé S.A., Vevey, Switzerland
| | - Tengfei Li
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Brittany R. Howell
- Fralin Biomedical Research Institute at VTC, Department of Human Development and Family Science, Virginia Polytechnic Institute and State University, Roanoke, VA, United States
| | - Kristine Baluyot
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Heather C. Hazlett
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jed T. Elison
- Institute of Child Development, University of Minnesota, Minneapolis, MN, United States
| | - Hongtu Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jonas Hauser
- Nestlé Institute of Health Sciences, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Norbert Sprenger
- Nestlé Institute of Health Sciences, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Weili Lin
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
29
|
Nuncio-Mora L, Lanzagorta N, Nicolini H, Sarmiento E, Ortiz G, Sosa F, Genis-Mendoza AD. The Role of the Microbiome in First Episode of Psychosis. Biomedicines 2023; 11:1770. [PMID: 37371865 DOI: 10.3390/biomedicines11061770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
The relationship between the gut-brain-microbiome axis has gained great importance in the study of psychiatric disorders, as it may represent a new target for their treatment. To date, the available literature suggests that the microbiota may influence the pathophysiology of several diseases, including psychosis. The aim of this review is to summarize the clinical and preclinical studies that have evaluated the differences in microbiota as well as the metabolic consequences related to psychosis. Current data suggest that the genera Lactobacillus and Megasphaera are increased in schizophrenia (SZ), as well as alterations in the glutamate-glutamine-GABA cycle, serum levels of tryptophan, kynurenic acid (KYNA), and short-chain fatty acids (SCFAs). There are still very few studies on early-onset psychosis, thus more studies are needed to be able to propose targeted therapies for a point when the disease has just started or has not yet progressed.
Collapse
Affiliation(s)
- Lucero Nuncio-Mora
- Laboratory of Genomics of Psychiatric and Neurodegenerative Diseases, National Institute of Genomic Medicine, Mexico City 14610, Mexico
- Posgraduate Studies in Biological Sciences, Posgraduate Unit, Posgraduate Circuit, Universitary City, Building D, 1st Floor, Coyoacan, Mexico City 04510, Mexico
| | | | - Humberto Nicolini
- Laboratory of Genomics of Psychiatric and Neurodegenerative Diseases, National Institute of Genomic Medicine, Mexico City 14610, Mexico
- Carraci Medical Group, Mexico City 03740, Mexico
| | - Emmanuel Sarmiento
- Psychiatric Children's Hospital Dr. Juan N. Navarro, Mexico City 14080, Mexico
| | - Galo Ortiz
- Psychiatric Children's Hospital Dr. Juan N. Navarro, Mexico City 14080, Mexico
| | - Fernanda Sosa
- Carraci Medical Group, Mexico City 03740, Mexico
- Psychiatric Children's Hospital Dr. Juan N. Navarro, Mexico City 14080, Mexico
| | - Alma Delia Genis-Mendoza
- Laboratory of Genomics of Psychiatric and Neurodegenerative Diseases, National Institute of Genomic Medicine, Mexico City 14610, Mexico
- Psychiatric Children's Hospital Dr. Juan N. Navarro, Mexico City 14080, Mexico
| |
Collapse
|
30
|
Gut microbial modulation by culinary herbs and spices. Food Chem 2023; 409:135286. [PMID: 36599291 DOI: 10.1016/j.foodchem.2022.135286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 11/30/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022]
Abstract
Culinary herbs and spices have previously been recognised for their potential impact on health through antioxidant and antimicrobial properties. They may also be promotors of positive microbial modulation by stimulating beneficial gut bacteria during fermentation, increasing the production of short chain fatty acids and thereby exhibiting a prebiotic effect. In the present paper, current literature around herb and spice consumption, gut microbiota modulation and prospective health benefits were reviewed. Herb and spice consumption can positively modulate gut microbes and possibly play an important role in inflammation related afflictions such as obesity. Current literature indicates that few human studies have been conducted to confirm the impact of herb and spice consumption on gut microbiota in connection with prospective health outcomes and inconsistencies in conclusions therefore remain.
Collapse
|
31
|
Hill DR, Buck RH. Infants Fed Breastmilk or 2'-FL Supplemented Formula Have Similar Systemic Levels of Microbiota-Derived Secondary Bile Acids. Nutrients 2023; 15:nu15102339. [PMID: 37242222 DOI: 10.3390/nu15102339] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/05/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Human milk represents an optimal source of nutrition during infancy. Milk also serves as a vehicle for the transfer of growth factors, commensal microbes, and prebiotic compounds to the immature gastrointestinal tract. These immunomodulatory and prebiotic functions of milk are increasingly appreciated as critical factors in the development of the infant gut and its associated microbial community. Advances in infant formula composition have sought to recapitulate some of the prebiotic and immunomodulatory functions of milk through human milk oligosaccharide (HMO) fortification, with the aim of promoting healthy development both within the gastrointestinal tract and systemically. Our objective was to investigate the effects of feeding formulas supplemented with the HMO 2'-fucosyllactose (2'-FL) on serum metabolite levels relative to breastfed infants. A prospective, randomized, double-blinded, controlled study of infant formulas (64.3 kcal/dL) fortified with varying levels of 2'-FL and galactooligosaccharides (GOS) was conducted [0.2 g/L 2'-FL + 2.2 g/L GOS; 1.0 g/L 2'-FL + 1.4 g/L GOS]. Healthy singleton infants age 0-5 days and with birth weight > 2490 g were enrolled (n = 201). Mothers chose to either exclusively formula-feed or breastfeed their infant from birth to 4 months of age. Blood samples were drawn from a subset of infants at 6 weeks of age (n = 35-40 per group). Plasma was evaluated by global metabolic profiling and compared to a breastfed reference group (HM) and a control formula (2.4 g/L GOS). Fortification of control infant formula with the HMO 2'-FL resulted in significant increases in serum metabolites derived from microbial activity in the gastrointestinal tract. Most notably, secondary bile acid production was broadly increased in a dose-dependent manner among infants receiving 2'-FL supplemented formula relative to the control formula. 2'-FL supplementation increased secondary bile acid production to levels associated with breastfeeding. Our data indicate that supplementation of infant formula with 2'-FL supports the production of secondary microbial metabolites at levels comparable to breastfed infants. Thus, dietary supplementation of HMO may have broad implications for the function of the gut microbiome in systemic metabolism. This trial was registered at with the U.S. National library of Medicine as NCT01808105.
Collapse
Affiliation(s)
- David R Hill
- Abbott, Nutrition Division, Columbus, OH 43219, USA
| | | |
Collapse
|
32
|
Devi MB, Sarma HK, Mukherjee AK, Khan MR. Mechanistic Insights into Immune-Microbiota Interactions and Preventive Role of Probiotics Against Autoimmune Diabetes Mellitus. Probiotics Antimicrob Proteins 2023:10.1007/s12602-023-10087-1. [PMID: 37171690 DOI: 10.1007/s12602-023-10087-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2023] [Indexed: 05/13/2023]
Abstract
Recent studies on genetically susceptible individuals and animal models revealed the potential role of the intestinal microbiota in the pathogenesis of type 1 diabetes (T1D) through complex interactions with the immune system. T1D incidence has been increasing exponentially with modern lifestyle altering normal microbiota composition, causing dysbiosis characterized by an imbalance in the gut microbial community. Dysbiosis has been suggested to be a potential contributing factor in T1D. Moreover, several studies have shown the potential role of probiotics in regulating T1D through various mechanisms. Current T1D therapies target curative measures; however, preventive therapeutics are yet to be proven. This review highlights immune microbiota interaction and the immense role of probiotics and postbiotics as important immunological interventions for reducing the risk of T1D.
Collapse
Affiliation(s)
- M Bidyarani Devi
- Molecular Biology and Microbial Biotechnology Laboratory, Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India
- Department of Biotechnology, Gauhati University, Guwahati, Assam, India
| | | | - Ashis K Mukherjee
- Molecular Biology and Microbial Biotechnology Laboratory, Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India
| | - Mojibur R Khan
- Molecular Biology and Microbial Biotechnology Laboratory, Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India.
| |
Collapse
|
33
|
Dou Y, Luo Y, Xing Y, Liu H, Chen B, Zhu L, Ma D, Zhu J. Human Milk Oligosaccharides Variation in Gestational Diabetes Mellitus Mothers. Nutrients 2023; 15:nu15061441. [PMID: 36986171 PMCID: PMC10059845 DOI: 10.3390/nu15061441] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/08/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is a common disease of pregnancy, but with very limited knowledge of its impact on human milk oligosaccharides (HMOs) in breast milk. This study aimed to explore the lactational changes in the concentration of HMOs in exclusively breastfeeding GDM mothers and the differences between GDM and healthy mothers. A total of 22 mothers (11 GDM mothers vs. 11 healthy mothers) and their offspring were enrolled in the study and the levels of 14 HMOs were measured in colostrum, transitional milk, and mature milk. Most of the HMOs showed a significant temporal trend with decreasing levels over lactation; however, there were some exceptions for 2′-Fucosyllactose (2′-FL), 3-Fucosyllactose (3-FL), Lacto-N-fucopentaose II (LNFP-II), and Lacto-N-fucopentaose III (LNFP-III). Lacto-N-neotetraose (LNnT) was significantly higher in GDM mothers in all time points and its concentrations in colostrum and transitional milk were correlated positively with the infant’s weight-for-age Z-score at six months postnatal in the GDM group. Significant group differences were also found in LNFP-II, 3′-Sialyllactose (3′-SL), and Disialyllacto-N-tetraose (DSLNT) but not in all lactational periods. The role of differently expressed HMOs in GDM needs to be further explored by follow-up studies.
Collapse
Affiliation(s)
- Yuqi Dou
- Department of Social Medicine and Health Education, School of Public Health, Peking University Health Science Center, Beijing 100191, China; (Y.D.)
| | - Yuanli Luo
- School of Public Health, Sichuan University, Chengdu 610041, China
| | - Yan Xing
- Department of Pediatrics, Peking University Third Hospital, Beijing 100191, China
| | - Hui Liu
- Department of Pediatrics, Peking University Third Hospital, Beijing 100191, China
| | - Botian Chen
- Department of Social Medicine and Health Education, School of Public Health, Peking University Health Science Center, Beijing 100191, China; (Y.D.)
| | - Liye Zhu
- Obstetrics Department, Maternal and Child Hospital of Haidian District, Beijing 100080, China
| | - Defu Ma
- Department of Social Medicine and Health Education, School of Public Health, Peking University Health Science Center, Beijing 100191, China; (Y.D.)
- Correspondence: (D.M.); (J.Z.)
| | - Jing Zhu
- Institute of Biotechnology and Health, Beijing Academy of Science and Technology, Beijing 100089, China
- Correspondence: (D.M.); (J.Z.)
| |
Collapse
|
34
|
Infant Fecal Fermentations with Galacto-Oligosaccharides and 2′-Fucosyllactose Show Differential Bifidobacterium longum Stimulation at Subspecies Level. CHILDREN 2023; 10:children10030430. [PMID: 36979988 PMCID: PMC10047592 DOI: 10.3390/children10030430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/26/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023]
Abstract
The objective of the current study was to evaluate the potential of 2′-FL and GOS, individually and combined, in beneficially modulating the microbial composition of infant and toddler (12–18 months) feces using the micro-Matrix bioreactor. In addition, the impacts of GOS and 2′-FL, individually and combined, on the outgrowth of fecal bifidobacteria at (sub)species level was investigated using the baby M-SHIME® model. For young toddlers, significant increases in the genera Bifidobacterium, Veillonella, and Streptococcus, and decreases in Enterobacteriaceae, Clostridium XIVa, and Roseburia were observed in all supplemented fermentations. In addition, GOS, and combinations of GOS and 2′-FL, increased Collinsella and decreased Salmonella, whereas 2′-FL, and combined GOS and 2′-FL, decreased Dorea. Alpha diversity increased significantly in infants with GOS and/or 2′-FL, as well as the relative abundances of the genera Veillonella and Akkermansia with 2′-FL, and Lactobacillus with GOS. Combinations of GOS and 2′-FL significantly stimulated Veillonella, Lactobacillus, Bifidobacterium, and Streptococcus. In all supplemented fermentations, Proteobacteria decreased, with the most profound decreases accomplished by the combination of GOS and 2′-FL. When zooming in on the different (sub)species of Bifidobacterium, GOS and 2’-FL were shown to be complementary in stimulating breast-fed infant-associated subspecies of Bifidobacterium longum in a dose-dependent manner: GOS stimulated Bifidobacterium longum subsp. longum, whereas 2′-FL supported outgrowth of Bifidobacterium longum subsp. infantis.
Collapse
|
35
|
Mills S, Yang B, Smith GJ, Stanton C, Ross RP. Efficacy of Bifidobacterium longum alone or in multi-strain probiotic formulations during early life and beyond. Gut Microbes 2023; 15:2186098. [PMID: 36896934 PMCID: PMC10012958 DOI: 10.1080/19490976.2023.2186098] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 02/24/2023] [Indexed: 03/11/2023] Open
Abstract
The significance of Bifidobacterium to human health can be appreciated from its early colonization of the neonatal gut, where Bifidobacterium longum represents the most abundant species. While its relative abundance declines with age, it is further reduced in several diseases. Research into the beneficial properties of B. longum has unveiled a range of mechanisms, including the production of bioactive molecules, such as short-chain fatty acids, polysaccharides, and serine protease inhibitors. From its intestinal niche, B. longum can have far-reaching effects in the body influencing immune responses in the lungs and even skin, as well as influencing brain activity. In this review, we present the biological and clinical impacts of this species on a range of human conditions beginning in neonatal life and beyond. The available scientific evidence reveals a strong rationale for continued research and further clinical trials that investigate the ability of B. longum to treat or prevent a range of diseases across the human lifespan.
Collapse
Affiliation(s)
- Susan Mills
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Bo Yang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | | | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Food Biosciences Department, Teagasc Food Research Centre, Co Cork, Ireland
| | - R. Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
36
|
Zenner C, Chalklen L, Adjei H, Dalby MJ, Mitra S, Cornwell E, Shaw AG, Sim K, Kroll JS, Hall LJ. Noninvasive Fecal Cytokine and Microbiota Profiles Predict Commencement of Necrotizing Enterocolitis in a Proof-of-Concept Study. GASTRO HEP ADVANCES 2023; 2:666-675. [PMID: 37469521 PMCID: PMC10352139 DOI: 10.1016/j.gastha.2023.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 03/01/2023] [Indexed: 07/21/2023]
Abstract
Background and Aims Necrotizing enterocolitis (NEC) is a life-threatening disease and the most common gastrointestinal emergency in premature infants. Accurate early diagnosis is challenging. Modified Bell's staging is routinely used to guide diagnosis, but early diagnostic signs are nonspecific, potentially leading to unobserved disease progression, which is problematic given the often rapid deterioration observed. We investigated fecal cytokine levels, coupled with gut microbiota profiles, as a noninvasive method to discover specific NEC-associated signatures that can be applied as potential diagnostic markers. Methods Premature babies born below 32 weeks of gestation were admitted to the 2-site neonatal intensive care unit (NICU) of Imperial College hospitals (St. Mary's or Queen Charlotte's & Chelsea) between January 2011 and December 2012. During the NICU stay, expert neonatologists grouped individuals by modified Bell's staging (healthy, NEC1, NEC2/3) and fecal samples from diapers were collected consecutively. Microbiota profiles were assessed by 16S rRNA gene amplicon sequencing and cytokine concentrations were measured by V-Plex multiplex assays. Results Early evaluation of microbiota profiles revealed only minor differences. However, at later time points, significant changes in microbiota composition were observed for Bacillota (adj. P = .0396), with Enterococcus being the least abundant in Bell stage 2/3 NEC. Evaluation of fecal cytokine levels revealed significantly higher concentrations of IL-1α (P = .045), IL-5 (P = .0074), and IL-10 (P = .032) in Bell stage 1 NEC compared to healthy individuals. Conclusion Differences in certain fecal cytokine profiles in patients with NEC indicate their potential use as diagnostic biomarkers to facilitate earlier diagnosis. Additionally, associations between microbial and cytokine profiles contribute to improving knowledge about NEC pathogenesis.
Collapse
Affiliation(s)
- Christian Zenner
- Intestinal Microbiome, School of Life Sciences, ZIEL – Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Lisa Chalklen
- Gut Microbes & Health, Quadram Institute Biosciences, Norwich, United Kingdom
| | - Helena Adjei
- Gut Microbes & Health, Quadram Institute Biosciences, Norwich, United Kingdom
| | - Matthew J. Dalby
- Gut Microbes & Health, Quadram Institute Biosciences, Norwich, United Kingdom
| | - Suparna Mitra
- Gut Microbes & Health, Quadram Institute Biosciences, Norwich, United Kingdom
- Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | - Emma Cornwell
- Department of Medicine, Section of Paediatric Infectious Disease, Imperial College London, London, United Kingdom
| | - Alexander G. Shaw
- Department of Medicine, Section of Paediatric Infectious Disease, Imperial College London, London, United Kingdom
| | - Kathleen Sim
- Department of Medicine, Section of Paediatric Infectious Disease, Imperial College London, London, United Kingdom
| | - J. Simon Kroll
- Department of Medicine, Section of Paediatric Infectious Disease, Imperial College London, London, United Kingdom
| | - Lindsay J. Hall
- Intestinal Microbiome, School of Life Sciences, ZIEL – Institute for Food & Health, Technical University of Munich, Freising, Germany
- Gut Microbes & Health, Quadram Institute Biosciences, Norwich, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|
37
|
de Freitas PNN, Silva CR, Constantin PP, Pileggi SAV, Vicari MR, Pileggi M. Fixing the Damage: The Evolution of Probiotics from Fermented Food to Biotherapeutic Products. A SUSTAINABLE GREEN FUTURE 2023:245-276. [DOI: 10.1007/978-3-031-24942-6_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
38
|
Asbjornsdottir B, Miranda-Ribera A, Fiorentino M, Konno T, Cetinbas M, Lan J, Sadreyev RI, Gudmundsson LS, Gottfredsson M, Lauth B, Birgisdottir BE, Fasano A. Prophylactic Effect of Bovine Colostrum on Intestinal Microbiota and Behavior in Wild-Type and Zonulin Transgenic Mice. Biomedicines 2022; 11:biomedicines11010091. [PMID: 36672598 PMCID: PMC9855927 DOI: 10.3390/biomedicines11010091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
The microbiota-gut-brain axis (MGBA) involves bidirectional communication between intestinal microbiota and the gastrointestinal (GI) tract, central nervous system (CNS), neuroendocrine/neuroimmune systems, hypothalamic-pituitary-adrenal (HPA) axis, and enteric nervous system (ENS). The intestinal microbiota can influence host physiology and pathology. Dysbiosis involves the loss of beneficial microbial input or signal, diversity, and expansion of pathobionts, which can lead to loss of barrier function and increased intestinal permeability (IP). Colostrum, the first milk from mammals after birth, is a natural source of nutrients and is rich in oligosaccharides, immunoglobulins, growth factors, and anti-microbial components. The aim of this study was to investigate if bovine colostrum (BC) administration might modulate intestinal microbiota and, in turn, behavior in two mouse models, wild-type (WT) and Zonulin transgenic (Ztm)-the latter of which is characterized by dysbiotic microbiota, increased intestinal permeability, and mild hyperactivity-and to compare with control mice. Bioinformatics analysis of the microbiome showed that consumption of BC was associated with increased taxonomy abundance (p = 0.001) and diversity (p = 0.004) of potentially beneficial species in WT mice and shifted dysbiotic microbial community towards eubiosis in Ztm mice (p = 0.001). BC induced an anxiolytic effect in WT female mice compared with WT female control mice (p = 0.0003), and it reduced anxiogenic behavior in Ztm female mice compared with WT female control mice (p = 0.001), as well as in Ztm male mice compared with WT BC male mice (p = 0.03). As evidenced in MGBA interactions, BC supplementation may well be applied for prophylactic approaches in the future. Further research is needed to explore human interdependencies between intestinal microbiota, including eubiosis and pathobionts, and neuroinflammation, and the potential value of BC for human use. The MGH Institutional Animal Care and Use Committee authorized the animal study (2013N000013).
Collapse
Affiliation(s)
- Birna Asbjornsdottir
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02152, USA
- School of Health Sciences, Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
- Unit for Nutrition Research, Landspitali University Hospital and Faculty of Food Science and Nutrition, University of Iceland, 101 Reykjavik, Iceland
- Correspondence:
| | - Alba Miranda-Ribera
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02152, USA
| | - Maria Fiorentino
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02152, USA
| | - Takumi Konno
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02152, USA
| | - Murat Cetinbas
- Department of Molecular Biology and Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jinggang Lan
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02152, USA
| | - Ruslan I. Sadreyev
- Department of Molecular Biology and Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Larus S. Gudmundsson
- School of Health Sciences, Faculty of Pharmaceutical Sciences, University of Iceland, 101 Reykjavik, Iceland
| | - Magnus Gottfredsson
- School of Health Sciences, Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
- Department of Scientific Affairs, Landspitali University Hospital, 101 Reykjavik, Iceland
- Department of Infectious Diseases, Landspitali University Hospital, 101 Reykjavik, Iceland
| | - Bertrand Lauth
- School of Health Sciences, Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
- Department of Child and Adolescent Psychiatry, Landspitali University Hospital, 105 Reykjavik, Iceland
| | - Bryndis Eva Birgisdottir
- Unit for Nutrition Research, Landspitali University Hospital and Faculty of Food Science and Nutrition, University of Iceland, 101 Reykjavik, Iceland
| | - Alessio Fasano
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02152, USA
- Department of Pediatrics, Harvard Medical School, Harvard University, Boston, MA 02114, USA
| |
Collapse
|
39
|
Food Peptides, Gut Microbiota Modulation, and Antihypertensive Effects. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248806. [PMID: 36557936 PMCID: PMC9788432 DOI: 10.3390/molecules27248806] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
The gut microbiota is increasingly important in the overall human health and as such, it is a target in the search of novel strategies for the management of metabolic disorders including blood pressure, and cardiovascular diseases. The link between microbiota and hypertension is complex and this review is intended to provide an overview of the mechanism including the production of postbiotics, mitigation of inflammation, and the integration of food biological molecules within this complex system. The focus is on hydrolyzed food proteins and peptides which are less commonly investigated for prebiotic properties. The analysis of available data showed that food peptides are multifunctional and can prevent gut dysbiosis by positively affecting the production of postbiotics or gut metabolites (short-chain fatty acids, polysaccharides, biogenic amines, bile acids). Peptides and the postbiotics then displayed antihypertensive effects via the renin-angiotensin system, the gut barrier, the endothelium, and reduction in inflammation and oxidative stress. Despite the promising antihypertensive effect of the food peptides via the modulation of the gut, there is a lack of human studies as most of the works have been conducted in animal models.
Collapse
|
40
|
Production of colanic acid hydrolysate and its use in the production of fucosylated oligosaccharides by engineered Saccharomyces cerevisiae. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.10.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
41
|
Álvarez-Mercado AI, Plaza-Diaz J. Dietary Polysaccharides as Modulators of the Gut Microbiota Ecosystem: An Update on Their Impact on Health. Nutrients 2022; 14:4116. [PMID: 36235768 PMCID: PMC9573424 DOI: 10.3390/nu14194116] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 09/30/2022] [Accepted: 10/01/2022] [Indexed: 12/13/2022] Open
Abstract
A polysaccharide is a macromolecule composed of more than ten monosaccharides with a wide distribution and high structural diversity and complexity in nature. Certain polysaccharides are immunomodulators and play key roles in the regulation of immune responses during the progression of some diseases. In addition to stimulating the growth of certain intestinal bacteria, polysaccharides may also promote health benefits by modulating the gut microbiota. In the last years, studies about the triad gut microbiota-polysaccharides-health have increased exponentially. In consequence, in the present review, we aim to summarize recent knowledge about the function of dietary polysaccharides on gut microbiota composition and how these effects affect host health.
Collapse
Affiliation(s)
- Ana I. Álvarez-Mercado
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Institute of Nutrition and Food Technology, Biomedical Research Center, University of Granada, 18016 Armilla, Spain
| | - Julio Plaza-Diaz
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
| |
Collapse
|
42
|
Góralczyk-Bińkowska A, Szmajda-Krygier D, Kozłowska E. The Microbiota-Gut-Brain Axis in Psychiatric Disorders. Int J Mol Sci 2022; 23:11245. [PMID: 36232548 PMCID: PMC9570195 DOI: 10.3390/ijms231911245] [Citation(s) in RCA: 173] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/10/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Modulating the gut microbiome and its influence on human health is the subject of intense research. The gut microbiota could be associated not only with gastroenterological diseases but also with psychiatric disorders. The importance of factors such as stress, mode of delivery, the role of probiotics, circadian clock system, diet, and occupational and environmental exposure in the relationship between the gut microbiota and brain function through bidirectional communication, described as "the microbiome-gut-brain axis", is especially underlined. In this review, we discuss the link between the intestinal microbiome and the brain and host response involving different pathways between the intestinal microbiota and the nervous system (e.g., neurotransmitters, endocrine system, immunological mechanisms, or bacterial metabolites). We review the microbiota alterations and their results in the development of psychiatric disorders, including major depressive disorder (MDD), schizophrenia (SCZ), bipolar disorder (BD), autism spectrum disorder (ASD), and attention-deficit hyperactivity disorder (ADHD).
Collapse
Affiliation(s)
- Aleksandra Góralczyk-Bińkowska
- Department of Microbiology and Experimental Immunology, MOLecoLAB: Lodz Centre of Molecular Studies on Civilisation Diseases, Medical University of Lodz, Mazowiecka 5 Street, 92-215 Lodz, Poland
| | - Dagmara Szmajda-Krygier
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1 Street, 90-151 Lodz, Poland
| | - Elżbieta Kozłowska
- Department of Microbiology and Experimental Immunology, MOLecoLAB: Lodz Centre of Molecular Studies on Civilisation Diseases, Medical University of Lodz, Mazowiecka 5 Street, 92-215 Lodz, Poland
| |
Collapse
|
43
|
The mechanism of probiotic action of human milk N-glycome towards B. infantis ATCC 15697 and identification of the principal functional components. Food Chem 2022; 384:132532. [DOI: 10.1016/j.foodchem.2022.132532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/14/2022] [Accepted: 02/19/2022] [Indexed: 11/18/2022]
|
44
|
Association of human milk oligosaccharides and nutritional status of young infants among Bangladeshi mother-infant dyads. Sci Rep 2022; 12:9456. [PMID: 35676397 PMCID: PMC9177541 DOI: 10.1038/s41598-022-13296-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
Human milk oligosaccharides (HMOs) support the development of a healthy gut microbiome and the growth of infants. We aimed to determine the association of different HMOs with severe acute malnutrition (SAM) among Bangladeshi young infants. This study was nested within a single-blind, randomized, pilot clinical trial (NCT0366657). A total of 45 breastmilk samples from mothers of < 6 months old infants who had SAM (n = 26) or were non-malnourished (n = 19) and were analyzed for constituent HMOs. Of the infants with SAM, 14 (53.85%) had secretor mothers, and 11 (57.89%) of the non-malnourished infants had secretor mothers. A one-unit increase in the relative abundance of sialylated HMOs was associated with higher odds of SAM in age and sex adjusted model (aOR = 2.00, 90% CI 1.30, 3.06), in age, sex, and secretor status adjusted model (aOR = 1.96, 90% CI 1.29, 2.98), and also in age and sex adjusted model among non-secretor mothers (aOR = 2.86, 90% CI 1.07, 7.62). In adjusted models, there was no evidence of a statistically significant association between SAM and fucosylated or undecorated HMOs. Our study demonstrates that a higher relative abundance of sialylated HMOs in mothers’ breastmilk may have a negative impact on young infants’ nutritional status.
Collapse
|
45
|
Leite JA, Robinson RC, Salcedo J, Ract JN, Quintal VS, Tadini CC, Barile D. The effect of microwave-assisted heating on bioactive and immunological compounds in donor human milk. Lebensm Wiss Technol 2022. [DOI: 10.1016/j.lwt.2022.113306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
46
|
Li Z, Zhu Y, Ni D, Zhang W, Mu W. Occurrence, functional properties, and preparation of 3-fucosyllactose, one of the smallest human milk oligosaccharides. Crit Rev Food Sci Nutr 2022; 63:9364-9378. [PMID: 35438024 DOI: 10.1080/10408398.2022.2064813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Human milk oligosaccharides (HMOs) are receiving wide interest and high attention due to their health benefits, especially for newborns. The HMOs-fortified products are expected to mimic human milk not only in the kinds of added oligosaccharides components but also the appropriate proportion between these components, and further provide the nutrition and physiological effects of human milk to newborns as closely as possible. In comparison to intensively studied 2'-fucosyllactose (2'-FL), 3-fucosyllactose (3-FL) has less attention in almost all respects. Nerveless, 3-FL naturally occurs in breast milk and increases roughly over the course of lactation with a nonnegligible content, and plays an irreplaceable role in human milk and delivers functional properties to newborns. According to the safety evaluation, 3-FL shows no acute oral toxicity, genetic toxicity, and subchronic toxicity. It has been approved as generally recognized as safe (GRAS). Biological production of 3-FL can be realized by enzymatic and cell factory approaches. The α1,3- or α1,3/4-fucosyltransferase is the key enzyme for 3-FL biosynthesis. Various metabolic engineering strategies have been applied to enhance 3-FL yield using cell factory approach. In conclusion, this review gives an overview of the recent scientific literatures regarding occurrence, bioactive properties, safety evaluation, and biotechnological preparation of 3-FL.
Collapse
Affiliation(s)
- Zeyu Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yingying Zhu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Dawei Ni
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wenli Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, China
| |
Collapse
|
47
|
Strain R, Stanton C, Ross RP. Effect of diet on pathogen performance in the microbiome. MICROBIOME RESEARCH REPORTS 2022; 1:13. [PMID: 38045644 PMCID: PMC10688830 DOI: 10.20517/mrr.2021.10] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/17/2022] [Accepted: 02/23/2022] [Indexed: 12/05/2023]
Abstract
Intricate interactions among commensal bacteria, dietary substrates and immune responses are central to defining microbiome community composition, which plays a key role in preventing enteric pathogen infection, a dynamic phenomenon referred to as colonisation resistance. However, the impact of diet on sculpting microbiota membership, and ultimately colonisation resistance has been overlooked. Furthermore, pathogens have evolved strategies to evade colonisation resistance and outcompete commensal microbiota by using unique nutrient utilisation pathways, by exploiting microbial metabolites as nutrient sources or by environmental cues to induce virulence gene expression. In this review, we will discuss the interplay between diet, microbiota and their associated metabolites, and how these can contribute to or preclude pathogen survival.
Collapse
Affiliation(s)
- Ronan Strain
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61 C996, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61 C996, Ireland
| | - R. Paul Ross
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Cork T12 YT20, Ireland
- School of Microbiology, University College Cork, College Road, Cork T12 K8AF, Ireland
| |
Collapse
|
48
|
Alliet P, Vandenplas Y, Roggero P, Jespers SNJ, Peeters S, Stalens JP, Kortman GAM, Amico M, Berger B, Sprenger N, Cercamondi CI, Corsello G. Safety and efficacy of a probiotic-containing infant formula supplemented with 2'-fucosyllactose: a double-blind randomized controlled trial. Nutr J 2022; 21:11. [PMID: 35193609 PMCID: PMC8862345 DOI: 10.1186/s12937-022-00764-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 02/08/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Human milk oligosaccharides (HMOs) have important and diverse biological functions in early life. This study tested the safety and efficacy of a starter infant formula containing Limosilactobacillus (L.) reuteri DSM 17938 and supplemented with 2'-fucosyllactose (2'FL). METHODS Healthy infants < 14 days old (n = 289) were randomly assigned to a bovine milk-based formula containing L. reuteri DSM 17938 at 1 × 107 CFU/g (control group; CG) or the same formula with added 1.0 g/L 2'FL (experimental group; EG) until 6 months of age. A non-randomized breastfed group served as reference (BF; n = 60). The primary endpoint was weight gain through 4 months of age in the formula-fed infants. Secondary endpoints included additional anthropometric measures, gastrointestinal tolerance, stooling characteristics, adverse events (AEs), fecal microbiota and metabolism, and gut immunity and health biomarkers in all feeding groups. RESULTS Weight gain in EG was non-inferior to CG as shown by a mean difference [95% CI] of 0.26 [-1.26, 1.79] g/day with the lower bound of the 95% CI above the non-inferiority margin (-3 g/day). Anthropometric Z-scores, parent-reported stooling characteristics, gastrointestinal symptoms and associated behaviors, and AEs were comparable between formula groups. Redundancy analysis indicated that the microbiota composition in EG was different from CG at age 2 (p = 0.050) and 3 months (p = 0.052), approaching BF. Similarly, between sample phylogenetic distance (weighted UniFrac) for BF vs EG was smaller than for BF vs CG at 3-month age (p = 0.045). At age 1 month, Clostridioides difficile counts were significantly lower in EG than CG. Bifidobacterium relative abundance in EG tracked towards that in BF. Fecal biomarkers and metabolic profile were comparable between CG and EG. CONCLUSION L. reuteri-containing infant formula with 2'FL supports age-appropriate growth, is well-tolerated and may play a role in shifting the gut microbial pattern towards that of breastfed infants. TRIAL REGISTRATION The trial was registered on ClinicalTrials.gov ( NCT03090360 ) on 24/03/2017.
Collapse
Affiliation(s)
| | - Yvan Vandenplas
- KidZ Health Castle, Vrije Universiteit Brussel, UZ Brussel, Brussels, Belgium
| | - Paola Roggero
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | | | | | | | | | - Mailis Amico
- Biostatistics & Data, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Bernard Berger
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Norbert Sprenger
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Colin I Cercamondi
- Nestlé Product Technology Center - Nutrition, Société des Produits Nestlé S.A., Vevey, Switzerland.
| | - Giovanni Corsello
- Department of Health Promotion Sciences Maternal and Infant Care, University of Palermo, Palermo, Italy
| |
Collapse
|
49
|
Chen G, Wu H, Zhu Y, Wan L, Zhang W, Mu W. Glycosyltransferase from Bacteroides gallinaceum Is a Novel α-1,3-Fucosyltransferase that Can Be Used for 3-Fucosyllactose Production In Vivo by Metabolically Engineered Escherichia coli. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:1934-1942. [PMID: 34989571 DOI: 10.1021/acs.jafc.1c06719] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
As one of the attractive fucosylated human milk oligosaccharides, the biological production of 3-fucosyllactose (3-FL) has received great attention, as it exhibits many excellent physiological functions for infants. In this work, a novel 3-FL-producing α-1,3-fucosyltransferase (α1,3-FucT) named FutM2 from Bacteroides gallinaceum was first selected from nine potential candidates in the NCBI database. Then, a highly 3-FL-producing engineered Escherichia coli strain was constructed by modular pathway enhancement including the GDL-l-fucose precursor supply by overexpressing manC, manB, gmd, and wcaG (CBGW), and the 3-FL synthesis pathway by introducing B. gallinaceum FutM2. Finally, a titer of 20.3 g L-1 and productivity of 0.40 g L-1 h-1 of 3-FL were achieved in the 3-L bioreactor by engineered E. coli (ΔlacZΔwcaJ) harboring pCDF-CBGW and pET-futM2. Our study provided a novel α1,3-FucT from B. gallinaceum that could be used for 3-FL production, presenting an efficient microbial cell factory platform to de novo synthesize 3-FL from glycerol.
Collapse
Affiliation(s)
- Geng Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Hao Wu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Bioengineering, Changsha University of Science & Technology, Changsha 410114, China
| | - Yingying Zhu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Li Wan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wenli Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wanmeng Mu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
50
|
De Cosmi V, Mazzocchi A, Agostoni C, Visioli F. Fructooligosaccharides: From Breast Milk Components to Potential Supplements. A Systematic Review. Adv Nutr 2022; 13:318-327. [PMID: 34555852 PMCID: PMC8803487 DOI: 10.1093/advances/nmab102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/12/2021] [Accepted: 08/25/2021] [Indexed: 12/30/2022] Open
Abstract
Breast milk is the optimal food choice for infant growth and development. Among breast milk components, fructooligosaccharides (FOSs) are being actively studied because of their role in microbiota development. In particular, 2'-fucosyllactose is being proposed as a potential supplement/nutraceutical or component of infant formula. In this systematic review, we critically summarize the available information on FOSs and we discuss their future use in infant nutrition. We searched the main electronic databases (PubMed, Embase, and Scopus), with a final check in May 2021. Search terms were inserted individually and using the Boolean tools AND and OR. Relevant articles were identified using the following words: ("fructooligosaccharides" OR "FOS") AND ("human milk" OR "breast milk" OR "donor milk" OR "bank milk"). The search retrieved 1814 articles. After removal of duplicates, we screened 1591 articles based on title, abstract, and exclusive use of the English language. We included articles describing the concentration of FOSs in human milk and assessed the relevant ones. We excluded reviews, studies on animals, and studies exclusively carried out on adults. Also, we excluded studies that have not reported evidence either on FOSs or on galactooligosaccharides from human milk. The resulting publications were reviewed, and 10 studies were included in the systematic review. We conclude that human milk FOSs are, indeed, crucial to infant gut development and their addition to infant formula is safe, well-tolerated, and might provide immune benefits to newborns. However, we would like to underscore the scantiness of human data and the need to avoid the immediate translation of infant research to the commercialization of supplements marketed to adults.
Collapse
Affiliation(s)
- Valentina De Cosmi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Alessandra Mazzocchi
- Pediatric Intermediate Care Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Carlo Agostoni
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy; Pediatric Intermediate Care Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Francesco Visioli
- Department of Molecular Medicine, University of Padua, Padua, Italy; Madrid Institute for Advanced Studies (IMDEA)-Food, Campus of International Excellence (CEI) Autonomous University of Madrid + Spanish National Research Council (UAM + CSIC), Madrid, Spain.
| |
Collapse
|